probiotics and inflammatory bowel disease · nutrition. on this basis, probiotics hold the...

26
1 Probiotics and Inflammatory Bowel Disease Gordon S. Howarth 1,2,3,4,5,6 1 Centre for Paediatric and Adolescent Gastroenterology, Children, Youth and Women’s Health Service; Disciplines of 2 Physiology, 3 Paediatrics, and 4 Agricultural and Animal Science, University of Adelaide; 5 School of Pharmacy and Biomedical Sciences, University of South Australia; 6 School of Biological Sciences, Flinders University, Adelaide, South Australia. Short Title: Probiotics and Inflammatory Bowel Disease. Correspondence: Associate Professor Gordon S. Howarth Centre for Paediatric and Adolescent Gastroenterology Children, Youth and Women’s Health Service 72 King William Road North Adelaide, South Australia, 5006, Australia Email: [email protected] Tel: 61-8-8161-6872 Fax: 61-8-8161-6088

Upload: others

Post on 26-Jan-2021

4 views

Category:

Documents


0 download

TRANSCRIPT

  • 1

    Probiotics and Inflammatory Bowel Disease

    Gordon S. Howarth1,2,3,4,5,6

    1Centre for Paediatric and Adolescent Gastroenterology, Children, Youth and Women’s Health

    Service; Disciplines of 2Physiology, 3Paediatrics, and 4Agricultural and Animal Science, University

    of Adelaide; 5School of Pharmacy and Biomedical Sciences, University of South Australia;

    6School of Biological Sciences, Flinders University, Adelaide, South Australia.

    Short Title: Probiotics and Inflammatory Bowel Disease.

    Correspondence: Associate Professor Gordon S. Howarth

    Centre for Paediatric and Adolescent Gastroenterology

    Children, Youth and Women’s Health Service

    72 King William Road

    North Adelaide, South Australia, 5006, Australia

    Email: [email protected]

    Tel: 61-8-8161-6872

    Fax: 61-8-8161-6088

    mailto:[email protected]

  • 2

    Abstract

    The aetiology of inflammatory bowel disease (IBD) remains obscure, currently thought to be

    associated with a genetic predisposition, dysregulation of the mucosal immune system, and a

    loss of antigen tolerance to enteric microflora, further influenced by a range of other

    environmental factors. In many cases, disease activity can be unremitting and refractory to

    treatment, with an unpredictable response to conventional therapy. To this end, new treatment

    strategies are being pursued on the basis of our understanding of IBD pathogenesis, and there

    is increasing evidence that at least some components of the enteric flora are primary

    contributors. Restoring the balance of the colonic microbiota to a less-pathogenic state is

    therefore a desirable strategy. Probiotics are currently defined as live non-pathogenic micro-

    organisms that, when ingested, exert a positive influence on host health beyond basic

    nutrition. On this basis, probiotics hold the potential to restore normal intestinal homeostasis.

    Despite more than a century of anecdotal reports of probiotic efficacy in gastrointestinal

    disease, only relatively recently have well-controlled, scientific studies and clinical trials,

    been conducted. Whilst reliable in vitro predictors of potential in vivo efficacy of putative

    probiotics await development, well-characterised animal model systems are proving valuable

    for the methodical, pre-clinical development of probiotics. Although early probiotic

    applications focussed largely on lactic acid bacteria (lactobacilli) and bifidobacteria, the

    range of candidate probiotics has now expanded significantly. Successful clinical application

    of the probiotic formulation, VSL#3, for treatment of the pouchitis variant of IBD, has

    instilled new excitement into the applicability of probiotics to IBD treatment, and the

    potential importance of probiotic combinations. The availability of new recombinant

    methodologies to develop ‘designer’ probiotics, capable of synthesizing and secreting specific

    factors, ranging from vitamins through to antibodies, further broadens the scope for probiotic

    application in IBD. Indeed, there are encouraging reports that probiotics may not need to be

    viable, or even intact, to exert their beneficial effects, with reports of therapeutic benefit from

    bacterial components such as DNA. In addition to the development of rigorous predictive

    systems to ascertain probiotic efficacy, challenges for the future will include determining the

  • 3

    optimal probiotic, or probiotic combination, and its timing of administration during phases of

    IBD relapse and remission. At present, our understanding of the intestinal microflora, and the

    importance of its composition and variability between individuals, is limited. However, once

    this understanding has been attained, strategically-designed probiotic formulations could

    ultimately be ‘tailored’ to suit individual IBD patients.

    Introduction

    Inflammatory bowel disease (IBD) is the collective term for a group of chronic, idiopathic

    inflammatory disorders that affect the gastrointestinal system. Crohn's disease and ulcerative

    colitis (UC) are the most common and serious variants of IBD, together imposing significant

    patient morbidity, economic burden, and long-term healthcare dependence. The aetiology of

    inflammatory bowel disease (IBD) remains obscure, currently thought to be associated with a

    genetic predisposition, dysregulation of the mucosal immune system, and a range of other

    environmental factors (1). In many cases, disease activity can be unremitting, with an

    unpredictable response to conventional therapy.

    Considerable progress has been made in studies of IBD genetics over the last decade, and the

    complementary strategies of genome-wide scanning and candidate gene-directed studies have

    led to the identification of a number of genetic markers that appear to predict disease

    susceptibility and behaviour (2). Identification of genetic markers that predispose to

    inflammation (alleles DR2, DRB1*0103 and DRB1*12) and a Crohn’s disease susceptibility

    gene on chromosome 16 [NOD2/CARD 15: Nucleotide-binding oligomerization domain

    protein 2] has paved the way for exciting new developments in therapeutic approaches for

    IBD (3,4). NOD2/CARD 15 is a cytosolic protein involved in intracellular recognition of

    microbes by sensing peptidoglycan fragments, and there is compelling evidence that it serves

    as an intracellular pattern recognition receptor to enhance host defence by inducing the

    production of antimicrobial peptides such as human beta-defensin-2 (5).

  • 4

    The characterisation of newly-identified NOD2/CARD-15 mutations is providing new

    information on the likely contribution of a defective host anti-microbial defence system to

    IBD aetiology and pathogenesis. This could have exciting implications for the use of

    candidate micro-organisms as a novel treatment strategy for IBD. Prospectively, this

    information would be particularly valuable when combined with a concomitant strategy to

    address the genetic basis of responsiveness to IBD therapy. Indeed, the spectrum of new

    treatment modalities for IBD has expanded exponentially in recent years (reviewed in 6).

    These genetic and genomic strategies could form the basis for better predicting the likelihood

    of responsiveness to newly-developed IBD therapies associated with the utilisation of micro-

    organisms, or factors derived from such organisms.

    Probiotics

    Although under continual review, the generally agreed definition of probiotics encompasses

    "live micro-organisms which, when consumed in adequate amounts, confer a health benefit

    on the host beyond basic nutrition" (7). This definition, however, may prove to be too limited

    and the term ‘alimentary pharmabiotics’ has been coined to further encompass dead

    organisms, or bacterial constituents which may be genetically or otherwise modified, and may

    not necessarily be restricted to those of human origin. Lactobacilli (8,9) and bifidobacteria,

    (10) species are generally referred to as archetypical probiotics, with increasing reports of

    probiotic properties attributed to non-pathogenic Escherichia coli (11,12) and non-bacterial

    organisms, such as Saccharomyces boulardii (13). Probiotics are thought to function through

    a number of different actions including: the production of antimicrobial agents such as

    bacteriocins, hydrogen peroxide and organic acids; blocking adhesion of pathogens or toxins

    to epithelial cells; providing antioxidant agents; and modulation of the immune system

    (14,15). Although the current review focuses on probiotic effects in the context of IBD, the

    complex interplay of mechanisms associated with specific organisms, or combinations of

    probiotic organisms, extends to applications for a broader range of medical disorders.

  • 5

    Probiotics and IBD

    Perhaps not surprisingly, the idiopathic nature of IBD, combined with its profound

    inflammatory characteristics, has resulted in current therapeutic strategies being targeted,

    almost exclusively, at disease immunomodulation (16). Notwithstanding this historical

    approach, new treatment modalities are now being pursued on the basis of our increased

    understanding of IBD pathogenesis. To this end, there is accumulating evidence that at least

    some components of the enteric flora are primary contributors to the unrelenting intestinal

    inflammation so characteristic of IBD. In general terms, restoring the balance of the colonic

    microbiota to a ‘less-pathogenic’ state would appear a desirable preventative, or therapeutic,

    strategy. However, before we consider probiotics as a potential therapeutic option in IBD, we

    should consider the resident enteric microflora and the potential impact of probiotic

    organisms in the context of IBD aetiology.

    The intestinal microbiota and IBD aetiology

    Although the intestinal flora is essential for host defence, some of its constituents may, in

    genetically susceptible hosts, become a risk factor for IBD, and it has been proposed that

    resident bacterial flora play a pivotal role in IBD pathogenesis (17). Mechanisms underlying

    the influence of the intestinal flora on mucosal homeostasis, mucosal protection, development

    and function of immune responses, and metabolism of fecal residue are undergoing increased

    scientific scrutiny. Strategies to enhance the beneficial properties of endogenous microflora,

    or alternatively, to minimise deleterious effects, represent a logical therapeutic approach,

    forming the basis for manipulation of the intestinal flora in IBD treatment (18).

    Nevertheless, it is important to understand that an inappropriate reaction to infection by a

    specific persistent pathogen has not yet been eliminated as a possible aetiological component

    in IBD. Indeed, Crohn’s disease and UC share histopathological similarities with defined

    intestinal infections, and occur in areas with highest luminal bacterial concentrations (17,18).

    Strengthened by the causative link between Helicobacter pylori and gastric ulceration (19),

    many microbial pathogens have been promoted as initiators and perpetuators of IBD. Perhaps

  • 6

    Mycobacterium avium, subspecies paratuberculosis (MAP) provides the most persuasive

    evidence as a causative agent in IBD since it manifests a Crohn’s-like phenotype in livestock

    in the condition known as Jonne’s disease (20). Other potential pathogenic causes have

    included M. kansaii, Escherichia coli, Diplostreptococcus species, viral vectors (measles,

    RNA viruses), Listeria monocytogenes, Fusobacterium necrophorum, Chlamydia species,

    Pseudomona maltophila and Helicobacter hepaticus (21,22,23). Taken together, identified

    probiotics capable of altering the intestinal environment such that colonization by the

    aforementioned species is inhibited, would therefore appear to be a logical strategy in IBD.

    Antibiotics have a defined role in the management of IBD and its complications, although

    their long-term usage is undesirable due to the risk of toxicity, bacterial resistance and

    overgrowth. Nevertheless, it would be fair to state that the current weight of scientific

    literature identifying a single organism as causative of IBD, although highly desirable, is not

    particularly compelling. Although several authorities, including the Joint Food and

    Agricultural Organization of the United Nations and the World Health Organization (24,25),

    have described selection criteria for probiotic organisms, to date, no reliable in vitro

    predictors of in vivo efficacy of putative probiotics have been identified. This has been

    compounded by the realization that individual probiotics do not appear to act through a single

    mechanism, further complicated by the likelihood that probiotic combinations, as opposed to

    specific candidates, may be indicated for certain disorders. Indeed, the mechanism of action

    of probiotics is likely to vary with different strains and may also be dependent on the clinical

    condition for which it is applied. Although well-characterised animal models are gaining

    momentum as predictive pre-clinical efficacy systems, a discussion of likely probiotic

    mechanisms is indicated.

    Mechanisms of probiotic action in IBD

    Although perhaps a little simplistic, the ability for probiotics to prevent or combat infections

    may be partially- or entirely dependent upon mutual competitive metabolic interactions with

  • 7

    potential pathogens, production of anti-microbial peptides such as bacteriocins (25) or

    inhibition of epithelial adherence and translocation by pathogens (26,27). Interestingly,

    Collado et al (28) reported that in general, bifidobacterial strains of animal origin

    demonstrated an improved capacity to inhibit and displace pathogens from human mucus than

    were human strains, further reporting that bifidobacteria were capable of producing

    antimicrobial peptides directed against Helicobacter pylori (29). Cross-species utilization of

    probiotics may therefore provide another important level of complexity for probiotic utility in

    clinical disorders such as IBD. In certain allergic disorders, including atopic eczema,

    probiotic influences on mucosal barrier function may be operative (30), whilst multiple

    mechanisms may account for anti-neoplastic effects (31).

    Probiotics, tolerance and the immune response

    Since control of bowel inflammation has generally been recognised as paramount in IBD, it is

    perhaps not surprising that down-regulation of mucosal inflammation has been identified as

    the primary focus of both conventional and probiotic-targeted therapy, fuelled by the

    cumulative clinical experience with anti-TNF based treatment successes (32).

    Rodents and humans are normally tolerant to autologous microbiota, and an association

    between breakdown of this tolerance and the development of chronic intestinal inflammation

    has been demonstrated (32). Potentially, pathological responses to components of the

    intestinal flora may occur under normal physiological conditions, but these may be

    suppressed by immunoregulatory mechanisms. In a recent review by Thompson-Chagoyán et

    al (33), 11 models of IBD have been described, in which inflammation was found to be

    dependent on the presence of normal flora; the absence of normal flora being associated with

    non-appearance of the condition (34,35,36,37). This phenomenon has been reported across

    species (mice, rats and guinea pigs), and to occur in manipulated organisms such as

    transgenic mice with targeted deletion of the T-cell receptor (TCRα), that spontaneously

    develop colitis in response to the gut microbiota (38). Mucosal inflammation in rats and mice

    with induced IBD has also been reported to respond to treatment with broad-spectrum

  • 8

    antibiotics (39). Moreover; in some animal model systems, colonisation with normal flora

    results in the rapid development of T-cell-mediated gut inflammation which can be

    transferred to other animals using activated T cells directed against enteric bacteria (40).

    Nevertheless, it appears that not all commensal bacteria have an equivalent ability to induce

    mucosal inflammation, which is also influenced by the host genetic background.

    Following cell surface recognition via Toll-like receptors (41), the anti-inflammatory effects

    of probiotics require signalling with the epithelium and hence, the mucosal immune system

    (42). Although transduction of bacterial signals into host immune responses presumably

    involves more than one pathway, NF-kappaB has been established as a central regulator of

    epithelial responses to invasive pathogens (43,44). Non-pathogenic components of the flora

    may attenuate pro-inflammatory responses by delaying degradation of IkappaB, the counter-

    regulatory factor to NF-kappaB (44). Indeed, it may be that probiotic bifidobacteria and

    lactobacilli may not use the same mechanism to achieve their anti-inflammatory effects as

    other signal transduction pathways begin to emerge.

    Clearly, a better understanding of the interplay between prokaryotes and eukaryotes, and

    subsequent effects on metabolic activity, will facilitate our knowledge of probiotic

    mechanisms. Bacterial adjuvants, including peptidoglycan, lipopolysaccharide, and DNA

    (CpG) bind to membrane-bound Toll-like receptors (TLR-2, 4, and 9. respectively), or

    cytoplasmic (NOD1 and NOD2) receptors (pattern recognition receptors), that activate

    nuclear factor-kappaB and transcription of many proinflammatory cytokines (45). Prokaryotic

    DNA perhaps represents the first description of a growing number of bacteria-sourced factors

    with the potential to alter host epithelial and mucosal immune responses. Un-methylated

    cytosine–guanine (CpG)-containing DNA, the ligand for Toll-like receptor 9 (TLR9), is a

    recently recognized microbial product with immuno-stimulatory and immuno-regulatory

    effects (46). TLR9 is expressed by many cell types located in the intestine, including

    epithelial cells and dendritic cells, and subcutaneous administration of immuno-stimulatory

  • 9

    DNA has been reported to reduce the severity of experimental and spontaneous colitis in

    murine models of IBD (47) via a mechanism attributed to TLR9 signalling (48).

    A decrease in the secretion of pro-inflammatory cytokines, IFN-gamma, TNF-alpha and IL-

    12, and interference with bacterial adherence to the epithelium has been demonstrated

    following probiotic administration, associated with NF-kappaB inhibition, heat-shock protein

    induction and proteasome inhibition, although NF-kappaB induction has also been

    demonstrated (49,50). Unexpectedly, many of these beneficial effects have been achieved not

    only by live bacteria, but also by gamma-irradiated non-viable bacteria, bacterial DNA

    components and probiotic-cultured media (49,51). Investigations into probiotic supernatants

    and their therapeutic potential in IBD are therefore forming the basis for new directions in

    IBD research. In summary, although mechanisms of probiotic action may vary, depending on

    the experimental or clinical context, and depending on differences in the host and in the

    bacterial strain, the engagement with host immunity is pivotal to probiotic action in IBD.

    Probiotics as therapeutic agents in IBD

    The human colon is a densely populated microbial ecosystem with several hundred bacterial

    species usually present with a total weight estimated to be several hundred grams (52). There

    are up to 1013–1014 total bacteria in the human intestinal tract, representing 10- to 20-fold

    more than the total number of tissue cells in the entire body, with most bacteria being obligate

    anaerobes, including clostridia, eubacteria, bacteroides groups and the genus bifidobacterium,

    such as Bifidobacterium bifidum and Bifidobacterium infantis (53).

    Probiotic efficacy has been confirmed in animal model studies by several investigators with

    different probiotic strains (37,53), although the use of probiotic therapy for IBD has only

    recently attracted serious interest from clinicians. Clinical trials in Crohn's disease with

    organisms, including lactobacillus (54) yeast (13), and coliforms (56), have been confounded

    by small patient numbers, differences in disease activity and variations in disease distribution

    (56). These deficiencies have been exacerbated by a growing, but poorly regulated,

  • 10

    commercial market for probiotics, often linked with somewhat tenuous, or exaggerated,

    claims for health benefits. Historically, the stability, optimal dose-range, frequency of

    administration and vehicle for delivery, have rarely been determined. However, at present in

    Europe, well-designed and appropriately controlled trials of individual probiotic preparations

    are underway in both Crohn's disease and ulcerative colitis, to ascertain their efficacy in both

    active disease and in prevention of relapse.

    Individual probiotics and IBD treatment

    The predominant, potentially health-enhancing, bacteria in IBD are the bifidobacteria and

    lactobacilli, both of which belong to the lactic acid bacteria (LAB) group (57). These two

    genera do not include any significant pathogenic species and their dominance in the faeces of

    breast-fed babies is thought to impart protection against infection (58,59). Most commercially

    available probiotics meet minimum selection criteria including acid and bile resistance and

    survival during gastrointestinal transit, but an ideal individual probiotic strain for any given

    indication has still yet to be defined.

    Lactobacilli and bifidobacteria have traditionally been the most common candidates for

    probiotic-based treatments in IBD. Although there is a growing body of scientific literature

    and a wealth of anecdotal information supporting the utility of lactobacillus strains as

    therapeutic agents in a range of alimentary disorders, progress has perhaps been hampered by

    poorly-controlled associations between probiotic administration and clearly-defined clinical

    end-points. Recently, Hawrelak et al (60) conducted a systematic review of six clinical trials

    investigating the capacity for Lactobacillus rhamnosus GG to prevent the onset of antibiotic-

    associated diarrhoea. As data sources, these investigators employed computer-based searches

    of MEDLINE, CINAHL, AMED, the Cochrane Controlled Trials Register and the Cochrane

    Database of Systematic Reviews and the bibliographies of relevant papers and previous meta-

    analyses. Four of the six trials found a significant reduction in the risk of antibiotic-associated

    diarrhoea with co-administration of Lactobacillus GG; one trial reported a reduced number of

  • 11

    days with antibiotic-induced diarrhoea, whilst the final trial found no benefit of Lactobacillus

    GG supplementation. These results are not unique in the context of retrospective probiotic

    studies, highlighting the need for additional research and the development of strictly-

    controlled predictive systems to clarify the effectiveness of Lactobacillus-based treatments

    beyond the effects of LGG in prevention of antibiotic-associated diarrhoea.

    Bifidobacterium, a member of the dominant microbiota (i.e. >108–109 colony forming unit

    (CFU)/g, represent up to 25% of the cultivable faecal bacteria in adults and 80% in infants.

    As probiotic agents, bifidobacteria have been studied for their efficacy in the prevention and

    treatment of a broad spectrum of animal and/or human gastrointestinal disorders, such as

    colonic transit disorders, intestinal infections, and colonic adenomas and cancer (10). Indeed,

    certain strains (eg Bifidobacterium animalis strain DN-173 010) have been reported to

    prevent or alleviate infectious diarrhoea through effects on the immune system and resistance

    to colonization by pathogens and there is some evidence that certain bifidobacteria may

    actually protect the host from pro-carcinogenic activity of intestinal flora (10).

    Non-pathogenic Escherichia coli, especially the Nissle 1917 strain, have generated increasing

    reports of efficacy in the context of inflammatory disorders. In ulcerative colitis, the non-

    pathogenic strain of E. coli Nissle demonstrates efficacy equivalent to that of mesalazine in

    ulcerative colitis (61,62). Kamada et al (63) utilising dextran sulphate sodium (DSS)-induced

    and IL-10 knock-out models of colitis have recently reported the non-pathogenic E. coli strain

    Nissle1917 to prevent both acute and chronic colitis, with its anti-inflammatory properties

    attributed not only to viable bacteria but also to heat-killed bacteria or its genomic DNA.

    Obermeier et al (64) demonstrated a pro-inflammatory effect of cytosin-guanosin dinucleotide

    (CpG)-oligodeoxynucleotide (ODN) treatment in established and chronic DSS-induced

    colitis, suggesting that DNA derived from luminal bacteria plays a role in the perpetuation of

    chronic intestinal inflammation. These investigators further speculated that treatment with

    adenoviral ODN (AV-ODN) could block the known CpG effects in IBD. The apparent

    discrepancy between bacterial DNA sources and effects on intestinal inflammation highlights

  • 12

    the need for well-controlled comparative studies of bacterial DNA, and, more specifically,

    CpG motifs, sourced from endogenous or probiotic bacteria.

    It should be noted, however, that probiotic properties have not been attributed solely to

    bacterial sources, since non-bacterial organisms, such as Saccharomyces boulardii, or even

    nematode parasites have been utilised for probiotic purposes (13,65) In summary, it seems

    unlikely that a single probiotic will be equally suited to all indications; selection of strains for

    disease-specific indications will be required, and it is in this capacity that well-conducted

    animal model studies will prove a valuable tool

    ‘Designer’ probiotics

    Genetic modification of food-grade commensal bacteria will need to be accommodated into

    our concept of probiotics. To this end, the term "pharmabiotic" has been developed as a more

    appropriate generic or umbrella term to encompass any form of therapeutic exploitation of the

    commensal flora (66,67). Food-grade bacteria can be modified, or engineered, to achieve

    specific functional activity. This can include delivery of anti-inflammatory cytokines or other

    biologically active molecules and vaccines to the gut. Of relevance to IBD, the food grade

    organism Lactococcus lactis, has been engineered to secrete IL-10 locally within the gut (68).

    When tested in two animal models of IBD, the magnitude of this effect was equivalent to

    corticosteroid therapy in its ability to decrease inflammation and disease severity. Another

    example of the potential applications of engineered commensal organisms is the genetic

    modification of lactobacilli resident within the female genital tract to express functional two-

    domain CD4 in order to confer protection against HIV infectivity in vitro (69). The future

    scope for this strategy is limitless, but public health and other safety concerns must be

    resolved before routine clinical use in humans can be instigated. Other examples of

    genetically modified (GM) microbes include the delivery of single-chain antibodies for

    pathogen-specific passive immunity (70,71) and bacterial-derived trefoil factors to promote

    healing and repair in the inflamed mouse gut (72).

  • 13

    Recently, Inglis et al (73) in a rodent model of intestinal mucositis, utilising the recently-

    developed non-invasive sucrose breath test, described a decrease in small intestinal

    inflammation following administration of the folic-acid secreting probiotic, Streptococcus

    thermophilus TH-4. Although not tested directly, these investigators speculated that the

    ‘trickle’-delivery of S. thermophilus TH-4 sourced folate to the intestinal enterocytes may

    have reduced the severity of intestinal damage induced by the folate depletion, and resulting

    inhibition of DNA synthesis, manifest by methotrexate-based chemotherapy. Similarly, Geier

    et al (74) have described a partial reduction of colonic inflammation in a rodent model system

    utilising Lactobacillus fermentum BR11 reportedly via its unique capacity to modify the

    cystine/cysteine equilibrium and hence, reduce oxidative stress.

    Public health concerns in relation to the release of genetically-modified organisms into the

    environment have replaced technological constraints as the major hurdles to be overcome with

    genetically-modified bacteria, and bio-containment has emerged as an environmental priority

    (75). Nevertheless, it would appear highly likely that future probiotic studies will be focussed

    not only on exploiting their existing beneficial properties, but also their capacity to deliver a

    broad range of specific factors, ranging from vitamins and antibodies through to strategically-

    developed, genetically-modified agonists and inhibitors.

    Probiotic mixtures and formulations

    Simple ingestion of a broad spectrum of probiotics would appear to be a pragmatic approach

    to cover a range of different indications and individual variations in host flora, although this

    would assume that individual probiotic constituents are not mutually antagonistic.

    Furthermore, it is a fundamental principle of therapeutic development that the properties and

    optimal usage of individual components of any mixture or formulation should be

    comprehensively determined before they can be recommended in combination. Undoubtedly,

    however, the most compelling evidence for probiotic efficacy in IBD has been reported with a

    combination of eight bacterial strains in the maintenance of remission of active ulcerative

  • 14

    colitis (76) and prevention of pouchitis (77). This combination, termed VSL#3, comprises

    four strains of lactobacilli and three strains of bifidobacteria, together with Streptococcus

    thermophilus. Indeed, there is now a sizeable scientific literature on VSL#3 and its

    application for a growing number of clinical disorders, with a recent report indicating

    hydrolytic activity against gliadin polypeptides suggesting a possible application for VSL#3

    in the control of coeliac disease (78).

    Rachmilewitz et al (48) have reported anti-inflammatory effects of VSL#3 in murine

    experimental colitis, mediated by Toll-like receptor (TLR9) signalling. Importantly, these

    authors described that intragastric and subcutaneous administration of probiotic DNA

    ameliorated the severity of experimental colitis, whereas methylated probiotic DNA, calf

    thymus DNA, and DNase-treated probiotics had no effect. Moreover, colitis severity was

    attenuated to the same extent by intragastric delivery of non-viable, gamma-irradiated, or

    viable probiotics, suggesting that the protective effects of probiotics are mediated by their

    own DNA rather than by their metabolites or ability to colonize the colon. The finding that

    live micro-organisms were not required to attenuate experimental colitis holds significant

    implications for the further development of probiotics as prophylactics or therapeutics in IBD.

    The development of further probiotic combinations for IBD treatment appears highly likely,

    although the current strategy will remain somewhat empirical until better-defined predictive

    in vivo and in vitro systems have been developed.

    The emergence of more compelling pre-clinical data from well-controlled animal model

    studies is therefore proving to be a positive strategy for probiotic efficacy studies in

    prospective IBD research. Such a strategy, in the absence of a rigorous, rapid throughput in

    vitro screening assay, will become vital given the likely complexity of probiotic formulations

    comprising mixtures of currently-characterised and newly-developed ‘designer’ probiotics.

    To test the matrix of probiotic formulations in human clinical trials, even if conducted

    strategically, would clearly be impossible.

  • 15

    Prebiotics and Synbiotics

    Prebiotics are defined as non-digestible food ingredients that beneficially affect the host by

    selectively stimulating the growth of bacterial species already established in the colon and

    thus improve host health (79). These are usually of a poly- or oligo-saccharide nature with

    studies largely confined to fructo-oligosaccharide (FOS), galacto-oligosaccharide (GOS) and

    maltodextrin (80). The health interest of the Bifidobacterium genus is reflected in the

    commonly-accepted definition of prebiotics: food ingredients that selectively stimulate the

    growth and activity of bacteria in the gut, usually bifidobacteria (bifidogenic effect) and

    lactobacilli, thereby producing health benefits.

    Although an exhaustive review of prebiotics is beyond the scope of this review, the

    importance of identified prebiotics in combination with dead or live probiotics (synbiotics), or

    biologically active bacterial metabolites, should not be under-stated. For example, Kanauchi

    et al (81) have recently described a synbiotic combination of germinated barley foodstuff

    (GBF) and Eubacterium limosum (E. limosum) and its potential as an adjunctive treatment for

    IBD. Although probiotic approaches for IBD include VSL#3, Nissle1917, Clostridium

    butyricum, and Bifidobacterium-fermented milk, Eubacteria have not been studied to any

    great extent. E. limosum is a commensal micro-organism that is promoted by GBF

    administration, and its metabolites include butyrate, which can accelerate intestinal epithelial

    growth and inhibit IL-6 production. GBF is therefore a prebiotic, and its unique

    characteristics make it highly suitable for applications in IBD. GBF prolongs remission in

    ulcerative colitis patients and also attenuates clinical activity in non-remissive colitic patients.

    The further complexity of incorporating the increasing development and number of prebiotics

    into the expanding range of newly-developed probiotic formulations described previously,

    greatly increases the number of synbiotic combinations which await testing in pre-clinical

    systems. However, although this enormous number of synbiotics and designer synbiotics may

    be intimidating from a pre-clinical efficacy perspective, there exists the increased potential to

    identify greater numbers of promising, efficacious candidate treatment formulations for IBD.

  • 16

    Combining this strategy with genetic and genomic approaches to define IBD susceptibility

    and probiotic responsiveness could ultimately result in individually-tailored probiotic-based

    treatment approaches in IBD.

    Personalised probiotics

    Optimal selection of a probiotic may even need to take into account individual variations in

    host diet and composition of gut flora. In this respect, the apparent influence of human genetic

    variability on intestinal bacterial composition is particularly intriguing (82). Furthermore, it

    seems unlikely that a single probiotic will be equally suited to all conditions. As a

    consequence, selection of strains for disease-specific indications will be required (27). It is

    beginning to become apparent that our definition of probiotics may be somewhat simplistic

    since there are indications a probiotic under certain circumstances, may not be a probiotic at

    all under altered physiological or pathogenic states, or even in different hosts. Different

    probiotics have distinct properties, and not all models of experimental colitis respond to the

    same probiotics (83). Almost certainly, there will not be a ‘one size fits all’ approach to

    probiotic use in IBD. Personalised, designer probiotics and synbiotics could therefore be a

    real therapeutic option for IBD sufferers in the future.

    Conclusions

    Probiotics continue to hold great promise for IBD treatment and prevention, but despite some

    significant advances, it would be fair to say that this field of research is still in its infancy.

    Understanding the mechanisms responsible for the beneficial effect of probiotics in

    inflammatory bowel disease and experimental colitis will aid our understanding of the role of

    endogenous and exogenous bacteria in IBD aetiology and pathogenesis. The finding that live

    probiotics may not be essential for therapeutic effects, and that these effects may also be

    obtained by the systemic route of administration, could have a major impact on the use and

    manufacture of probiotics. Phenotypic and genomic characterisation of probiotic strains will

    be required, together with clarification of their mechanisms of action across a broad range of

  • 17

    rigorously-controlled clinical settings. Moreover, results of probiotic studies demonstrating

    efficacy in a given clinical setting should not be extrapolated to other disease indications

    without separate controlled assessments.

    Over the past decade, there have been quantum advances in probiotic-based research in IBD.

    Initial, largely anecdotal, reports of lactobacillus efficacy have been replaced by the

    development of specific ‘designer’ probiotics, which together with identified prebiotics, could

    result in a significant repertoire of designer synbiotics for IBD treatment. Such an array of

    treatment options, combined with genetic advances in IBD susceptibility and probiotic

    responsiveness, could eventually result in the exciting emergence of individually-tailored

    probiotic-sourced formulations. The major challenge in the short-term will be the

    development of rigorous in vivo and in vitro testing systems to rapidly determine the

    suitability of any given probiotic-based formulation, not only to a specific condition, but also

    to the individual.

    Acknowledgements

    The author would like to thank Associate Professor Ross Butler for his inspirational

    contributions into the preparation of this review, Mr Mark Geier for his valued comments and

    critique, and Miss Kerry Jolly for editorial assistance.

    References

    1. Schmidt C, Stallmach A. Etiology and pathogenesis of inflammatory bowel disease.

    Minerva Gastroenterol Dietol. 2005;51(2):127-45.

    2. Vermeire S, Rutgeerts P. Current status of genetics research in inflammatory bowel

    disease. Genes Immun. 2005;6(8):637-45.

    3. Ogura Y, Inohara N, Benito A, Chen FF, Yamaoka S, Nunez G. Nod2, a Nod1/Apaf-

    1 family member that is restricted to monocytes and activates NF-kappaB. J Biol

    Chem. 2001;276(7):4812-8.

  • 18

    4. Hampe J, Grebe J, Nikolaus S, Solberg C, Croucher PJ, Mascheretti S, Jahnsen J,

    Moum B, Klump B, Krawczak M, Mirza MM, Foelsch UR, Vatn M, Schreiber S.

    Association of NOD2 (CARD 15) genotype with clinical course of Crohn's disease: a

    cohort study. Lancet. 2002;359(9318):1661-5.

    5. Voss E, Wehkamp J, Wehkamp K, Stange EF, Schroder JM, Harder J.

    NOD2/CARD15 mediates induction of the antimicrobial peptide human beta-

    defensin-2. J Biol Chem. 2005 Nov 30; [Epub ahead of print].

    6. Isaacs KL, Lewis JD, Sandborn WJ, Sands BE, Targan SR. State of the art: IBD

    therapy and clinical trials in IBD. Inflamm Bowel Dis. 2005;Suppl 1:S3-12.

    7. Schrezenmeir J, de Vrese M. Probiotics, prebiotics, and synbiotics--approaching a

    definition. Am J Clin Nutr. 2001; (Suppl):361S-364S.

    8. Sartor RB. Probiotic therapy of intestinal inflammation and infections. Curr Opin

    Gastroenterol. 2005;21(1):44-50.

    9. Herias MV, Koninkx JF, Vos JG, Huis in't Veld JH, van Dijk JE. Probiotic effects of

    Lactobacillus casei on DSS-induced ulcerative colitis in mice. Int J Food Microbiol.

    2005;103(2):143-55.

    10. Picard C, Fioramonti J, Francois A, Robinson T, Neant F, Matuchansky C. Review

    article: bifidobacteria as probiotic agents -- physiological effects and clinical benefits.

    Aliment Pharmacol Ther. 2005;22(6):495-512.

    11. Kruis, P Fric¡, J Pokrotnieks, M Luka´s¡, B Fixa, M Kas¡c¡a´k, M A Kamm, J

    Weismueller, C Beglinger, M Stolte, C Wolff, J Schulze. Maintaining remission of

    ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as

    with standard mesalazine. Gut 2004;53:1617–1623.

    12. Schultz M, Strauch UG, Linde HJ, Watzl S, Obermeier F, Gottl C, Dunger N,

    Grunwald N, Scholmerich J, Rath HC. Preventive effects of Escherichia coli strain

    Nissle 1917 on acute and chronic intestinal inflammation in two different murine

    models of colitis. Clin Diagn Lab Immunol. 2004;11(2):372-8.

  • 19

    13. Guslandi M, Mezzi G, Sorghi M, Testoni PA. Saccharomyces boulardii in

    maintenance treatment of Crohn's disease. Dig Dis Sci. 2000;45(7):1462-4.

    14. Shanahan F. Probiotics and inflammatory bowel disease: from fads and fantasy to

    facts and future. Br J Nutr. 2002;88 Suppl 1:S5-9.

    15. Marteau P, Shanahan F. Basic aspects and pharmacology of probiotics: an overview

    of pharmacokinetics, mechanisms of action and side-effects. Best Pract Res Clin

    Gastroenterol. 2003;17(5):725-40.

    16. Shanahan F. Probiotics and the immune response: how much can we expect? J

    Pediatr Gastroenterol Nutr. 2004;39 Suppl 3:S748-9.

    17. Shanahan F. Host-flora interactions in inflammatory bowel disease. Inflamm Bowel

    Dis. 2004;10 Suppl 1:S16-24.

    18. Shanahan F. Probiotics in inflammatory bowel disease—therapeutic rationale and

    role. Adv Drug Deliv Rev. 2004;56(6):809-18.

    19. Marshall BJ, Warren JR. Unidentified curved bacilli in the stomach of patients with

    gastritis and peptic ulceration. Lancet. 1984;16;1(8390):1311-5.

    20. Manning EJ. Mycobacterium avium subspecies paratuberculosis: a review of current

    knowledge. J Zoo Wildl Med. 2001;32(3):293-304.

    21. Farrell R.J., La Mont J.T. Microbial factors in inflammatory bowel disease,

    Gastroenterol Clin North Am 2002;31 (1):41–62.

    22. McKay DM. Intestinal inflammation and the gut microflora. Can J Gastroenterol.

    1999;13(6):509-16.

    23. Report of a Joint F.A.O./W.H.O. Expert Consultation. Health and Nutritional

    Properties of Probiotics in Food Including Powder Milk and Live Lactic Acid

    Bacteria. http://www.fao.org/es/ESN/Probio/report, 2001.

    24. Working Group Report. Guidelines for the evaluation of probiotics in food. Joint

    Food and Agriculture Organization of the United Nations and The World Health

    Organization, London, Ontario, Canada April 30 and May 1, 2002.

  • 20

    25. Flynn S, van Sinderen D, Thornton GM, Holo H, Nes IF, Collins JK. Characterization

    of the genetic locus responsible for the production of ABP-118, a novel bacteriocin

    produced by the probiotic bacterium Lactobacillus salivarius subsp. salivarius

    UCC118. Microbiology. 2002;148(Pt 4):973-84.

    26. Bengmark S. Ecological control of the gastrointestinal tract. The role of probiotic

    flora. Gut.1998;42:2–7.

    27. Shanahan F. Probiotics in inflammatory bowel disease: is there a scientific rationale?.

    Inflamm. Bowel Dis. 2000;6:107–1115.

    28. Collado MC, Gueimonde M, Hernandez M, Sanz Y, Salminen S. Adhesion of

    selected Bifidobacterium strains to human intestinal mucus and the role of adhesion

    in enteropathogen exclusion. J Food Prot. 2005;68(12):2672-8.

    29. Collado MC, Gonzalez A, Gonzalez R, Hernandez M, Ferrus MA, Sanz Y.

    Antimicrobial peptides are among the antagonistic metabolites produced by

    Bifidobacterium against Helicobacter pylori. Int J Antimicrob Agents.

    2005;25(5):385-91.

    30. Isolauri E, Majamaa H, Arvola T, Rantala I, Virtanen E, Arvilommi H. Lactobacillus

    casei strain GG reverses increased intestinal permeability induced by cow milk in

    suckling rats. Gastroenterology. 1993;105(6):1643-1650.

    31. Dugas B, Mercenier A, Lenoir-Wijnkoop I, Arnaud C, Dugas N, Postaire E.

    Immunity and probiotics. Immunol Today. 1999;20(9):387-90.

    32. Sandborn WJ. New concepts in anti-tumor necrosis factor therapy for inflammatory

    bowel disease. Rev Gastroenterol Disord. 2005;5(1):10-8.

    33. Thompson-Chagoyan OC, Maldonado J, Gil A. Aetiology of inflammatory bowel

    disease (IBD): role of intestinal microbiota and gut-associated lymphoid tissue

    immune response. Clin Nutr. 2005;24(3):339-52.

    34. Teitelbaum JE, Walker WA. Nutritional impact of pre- and probiotics as protective

    gastrointestinal organisms. Annu Rev Nutr. 2002;22:107-38.

  • 21

    35. Taurog JD, Richardson JA, Croft JT, Simmons WA, Zhou M, Fernandez-Sueiro JL,

    Balish E, Hammer RE. The germfree state prevents development of gut and joint

    inflammatory disease in HLA-B27 transgenic rats. J Exp Med. 1994;180(6):2359-64.

    36. Sellon RK, Tonkonogy S, Schultz M, Dieleman LA, Grenther W, Balish E, Rennick

    DM, Sartor RB. Resident enteric bacteria are necessary for development of

    spontaneous colitis and immune system activation in interleukin-10-deficient mice.

    Infect Immun. 1998;66(11):5224-31.

    37. Madsen KL, Doyle JS, Jewell LD, Tavernini MM, Fedorak RN. Lactobacillus species

    prevents colitis in interleukin 10 gene-deficient mice. Gastroenterology.

    1999;116(5):1107-14.

    38. Mombaerts P, Mizoguchi E, Grusby MJ, Glimcher LH, Bhan AK, Tonegawa S.

    Spontaneous development of inflammatory bowel disease in T cell receptor mutant

    mice. Cell. 1993;75(2):274-82.

    39. Videla S, Vilaseca J, Guarner F, Salas A, Treserra F, Crespo E, Antolin M,

    Malagelada JR. Role of intestinal microflora in chronic inflammation and ulceration

    of the rat colon. Gut. 1994;35(8):1090-7.

    40. Aranda R, Sydora BC, McAllister PL, Binder SW, Yang HY, Targan SR, Kronenberg

    M. Analysis of intestinal lymphocytes in mouse colitis mediated by transfer of CD4+,

    CD45RBhigh T cells to SCID recipients. J Immunol. 1997;158(7):3464-73.

    41. Heine H, Ulmer AJ. Recognition of bacterial products by toll-like receptors. Chem

    Immunol Allergy. 2005;86:99-119.

    42. Neish AS, Gewirtz AT, Zeng H, Young AN, Hobert ME, Karmali V, Rao AS,

    Madara JL. Prokaryotic regulation of epithelial responses by inhibition of IkappaB-

    alpha ubiquitination. Science. 2000;289(5484):1560-3.

    43. Elewaut D, DiDonato JA, Kim JM, Truong F, Eckmann L, Kagnoff MF. NF-kappa B

    is a central regulator of the intestinal epithelial cell innate immune response induced

    by infection with enteroinvasive bacteria. J Immunol. 1999;163(3):1457-66.

  • 22

    44. Gewirtz AT, Rao AS, Simon PO Jr, Merlin D, Carnes D, Madara JL, Neish AS.

    Salmonella typhimurium induces epithelial IL-8 expression via Ca(2+)-mediated

    activation of the NF-kappaB pathway. J Clin Invest. 2000;105(1):79-92.

    45. Sartor RB. Targeting enteric bacteria in treatment of inflammatory bowel diseases:

    why, how, and when. Curr Opin Gastroenterol. 2003 Jul;19(4):358-65.

    46. Watson JL, McKay DM. The immunophysiological impact of bacterial CpG DNA on

    the gut. Clin Chim Acta. 2006;364(1-2):1-11.

    47. Rachmilewitz D, Karmeli F, Takabayashi K, Hayashi T, Leider-Trejo L, Lee J, Leoni

    LM, Raz E. Immunostimulatory DNA ameliorates experimental and spontaneous

    murine colitis. Gastroenterology. 2002;122(5):1428-41.

    48. Rachmilewitz D, Katakura K, Karmeli F, Hayashi T, Reinus C, Rudensky B, Akira S,

    Takeda K, Lee J, Takabayashi K, Raz E. Toll-like receptor 9 signaling mediates the

    anti-inflammatory effects of probiotics in murine experimental colitis.

    Gastroenterology. 2004;126(2):520-8.

    49. Dotan I, Rachmilewitz D. Probiotics in inflammatory bowel disease: possible

    mechanisms of action. Curr Opin Gastroenterol. 2005;21(4):426-30.

    50. Petrof EO, Kojima K, Ropeleski MJ, Musch MW, Tao Y, De Simone C, Chang EB.

    Probiotics inhibit nuclear factor-kappaB and induce heat shock proteins in colonic

    epithelial cells through proteasome inhibition. Gastroenterology. 2004;127(5):1474-

    87.

    51. Jijon H, Backer J, Diaz H, Yeung H, Thiel D, McKaigney C, De Simone C, Madsen

    K. DNA from probiotic bacteria modulates murine and human epithelial and immune

    function. Gastroenterology. 2004;126(5):1358-73.

    52. Bocci V. The neglected organ: bacterial flora has a crucial immunostimulatory role.

    Perspect Biol Med. 1992;35(2):251-60.

    53. O'Mahony L, Feeney M, O'Halloran S, Murphy L, Kiely B, Fitzgibbon J, Lee G,

    O'Sullivan G, Shanahan F, Collins JK. Probiotic impact on microbial flora,

  • 23

    inflammation and tumour development in IL-10 knockout mice. Aliment Pharmacol

    Ther. 2001;15(8):1219-25.

    54. Prantera C, Scribano ML, Falasco G, Andreoli A, Luzi C. Ineffectiveness of

    probiotics in preventing recurrence after curative resection for Crohn's disease: a

    randomised controlled trial with Lactobacillus GG. Gut. 2002;51(3):405-9.

    55. Malchow HA. Crohn's disease and Escherichia coli. A new approach in therapy to

    maintain remission of colonic Crohn's disease? J Clin Gastroenterol. 1997;25(4):653-

    8.

    56. Kruis W, Schutz E, Fric P, Fixa B, Judmaier G, Stolte M. Double-blind comparison

    of an oral Escherichia coli preparation and mesalazine in maintaining remission of

    ulcerative colitis. Aliment Pharmacol Ther. 1997;11(5):853-8.

    57. Salminen S, Bouley C, Boutron-Ruault M-C, et al. Functional food science and

    gastrointestinal physiology and function. British Journal of Nutrition 1998;80(Suppl.

    1): S147–71.

    58. Yoshioka H, Isaki K, Fujita K. Development and differences of intestinal flora in the

    neonatal period in breast-fed and bottle-fed infants. Pediatrics. 1983;72: 317–21.

    59. Harmsen HJ, Wildeboer-Veloo AC, Raangs GC, et al. Analysis of intestinal flora

    development in breast-fed and formula-fed infants by using molecular identification

    and detection methods. J Pediatr Gastroenterol Nutr. 2000;30: 61–7.

    60. Hawrelak JA, Whitten DL, Myers SP. Is Lactobacillus rhamnosus GG effective in

    preventing the onset of antibiotic-associated diarrhoea?: a systematic review.

    Digestion. 2005;72(1):51-6.

    61. Rembacken BJ, Snelling AM, Hawkey PM, Chalmers DM, Axon AT. Non-

    pathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis:

    a randomised trial. Lancet. 1999;354(9179):635-9.

    62. Gionchetti P, Rizzello F, Venturi A, Brigidi P, Matteuzzi D, Bazzocchi G, Poggioli

    G, Miglioli M, Campieri M. Oral bacteriotherapy as maintenance treatment in

  • 24

    patients with chronic pouchitis: a double-blind, placebo-controlled trial.

    Gastroenterology. 2000;119(2):305-9.

    63. Kamada N, Inoue N, Hisamatsu T, Okamoto S, Matsuoka K, Sato T, Chinen H, Hong

    KS, Yamada T, Suzuki Y, Suzuki T, Watanabe N, Tsuchimoto K, Hibi T. Non-

    pathogenic Escherichia coli strain Nissle1917 prevents murine acute and chronic

    colitis. Inflamm Bowel Dis. 2005;11(5):455-63.

    64. Obermeier F, Dunger N, Strauch UG, Hofmann C, Bleich A, Grunwald N, Hedrich

    HJ, Aschenbrenner E, Schlegelberger B, Rogler G, Scholmerich J, Falk W. CpG

    motifs of bacterial DNA essentially contribute to the perpetuation of chronic

    intestinal inflammation. Gastroenterology. 2005;129(3):913-27.

    65. Summers RW, Elliott DE, Urban JF Jr, Thompson RA, Weinstock JV. Trichuris

    therapy for active ulcerative colitis: a randomized controlled trial. Gastroenterology.

    2005;128(4):825-32.

    66. Shanahan F. Physiological basis for novel drug therapies used to treat the

    inflammatory bowel diseases I. Pathophysiological basis and prospects for probiotic

    therapy in inflammatory bowel disease. Am J Physiol Gastrointest Liver Physiol.

    2005;288(3):G417-21.

    67. O'Sullivan GC, Kelly P, O'Halloran S, Collins C, Collins JK, Dunne C, Shanahan F.

    Probiotics: an emerging therapy. Curr Pharm Des. 2005;11(1):3-10.

    68. Huyghebaert N, Vermeire A, Rottiers P, Remaut E, Remon JP. Development of an

    enteric-coated, layered multi-particulate formulation for ileal delivery of viable

    recombinant Lactococcus lactis. Eur J Pharm Biopharm. 2005;61(3):134-41.

    69. Chang TL, Chang CH, Simpson DA, Xu Q, Martin PK, Lagenaur LA, Schoolnik GK,

    Ho DD, Hillier SL, Holodniy M, Lewicki JA, Lee PP. Inhibition of HIV infectivity

    by a natural human isolate of Lactobacillus jensenii engineered to express functional

    two-domain CD4. Proc Natl Acad Sci U S A. 2003 Sep 30;100(20):11672-7.

    70. Beninati C, Oggioni MR, Boccanera M, Spinosa MR, Maggi T, Conti S, Magliani W,

    De Bernardis F, Teti G, Cassone A, Pozzi G, Polonelli L. Therapy of mucosal

  • 25

    candidiasis by expression of an anti-idiotype in human commensal bacteria. Nat

    Biotechnol. 2000;18(10):1060-4.

    71. Kruger C, Hu Y, Pan Q, Marcotte H, Hultberg A, Delwar D, van Dalen PJ, Pouwels

    PH, Leer RJ, Kelly CG, van Dollenweerd C, Ma JK, Hammarstrom L. In situ

    delivery of passive immunity by lactobacilli producing single-chain antibodies. Nat

    Biotechnol. 2002;20(7):702-6.

    72. Vandenbroucke K, Hans W, Van Huysse J, Neirynck S, Demetter P, Remaut E,

    Rottiers P, Steidler L. Active delivery of trefoil factors by genetically modified

    Lactococcus lactis prevents and heals acute colitis in mice. Gastroenterology.

    2004;127(2):502-13.

    73. Inglis KL, Howarth GS, Davidson GP, Butler RN. Oral ingestion of Streptococcus

    thermophilus diminishes severity of small intestinal mucositis in methotrexate

    treated rats Cancer Biol Therap. 2006. (In Press).

    74. Geier MS, Butler RN, Giffard PM, Howarth GS. Differential Efficacy of Probiotic

    Species in Amelioration of Dextran Sulphate Sodium-Induced Colitis in Rats.

    Gastroenterology. 2005;128 (4): A-603.

    75. Hampton T. Prevent genetically modified organisms from escaping into nature, report

    urges. JAMA. 2004;291(9):1055.

    76. Bibiloni R, Fedorak RN, Tannock GW, Madsen KL, Gionchetti P, Campieri M, De

    Simone C, Sartor RB. VSL#3 probiotic-mixture induces remission in patients with

    active ulcerative colitis. Am J Gastroenterol. 2005;100(7):1539-46.

    77. Lammers KM, Vergopoulos A, Babel N, Gionchetti P, Rizzello F, Morselli C,

    Caramelli E, Fiorentino M, d'Errico A, Volk HD, Campieri M. Probiotic therapy in

    the prevention of pouchitis onset: decreased interleukin-1beta, interleukin-8, and

    interferon-gamma gene expression. Inflamm Bowel Dis. 2005;11(5):447-54.

    78. Angelis MD, Rizzello CG, Fasano A, Clemente MG, Simone CD, Silano M, Vi

    ncenzi MD, Losito I, Gobbetti M. VSL#3 probiotic preparation has the capacity to

  • 26

    hydrolyze gliadin polypeptides responsible for Celiac Sprue probiotics and gluten

    intolerance. Biochim Biophys Acta. 2005;1762(1):80-93.

    79. Gibson GR. Dietary modulation of the human gut microflora using prebiotics. Br J

    Nutr. 1998;80(4):S209-12.

    80. Rastall RA, Gibson GR, Gill HS, Guarner F, Klaenhammer TR, Pot B, Reid G,

    Rowland IR, Sanders ME. Modulation of the microbial ecology of the human colon

    by probiotics, prebiotics and synbiotics to enhance human health: an overview of

    enabling science and potential applications. FEMS Microbiol Ecol. 2005;52(2):145-

    52.

    81. Kanauchi O, Mitsuyama K, Araki Y, Andoh A. Modification of intestinal flora in the

    treatment of inflammatory bowel disease. Curr Pharm Des. 2003;9(4):333-46.

    82. Van de Merwe JP, Stegeman JH, Hazenberg MP.The resident faecal flora is

    determined by genetic characteristics of the host. Implications for Crohn's disease?

    Antonie Van Leeuwenhoek. 1983;49(2):119-24.

    83. Fedorak RN, Madsen KL. Probiotics and prebiotics in gastrointestinal disorders. Curr

    Opin Gastroenterol. 2004;20(2):146-55.

    Probiotics and Inflammatory Bowel DiseaseShort Title: Probiotics and Inflammatory Bowel Disease. Probiotics and IBD The intestinal microbiota and IBD aetiologyMechanisms of probiotic action in IBDAlthough perhaps a little simplistic, the ability for probiotics to prevent or combat infections may be partially- or entirely dependent upon mutual competitive metabolic interactions with potential pathogens, production of anti-microbial peptides such as bacteriocins (25) or inhibition of epithelial adherence and translocation by pathogens (26,27). Interestingly, Collado et al (28) reported that in general, bifidobacterial strains of animal origin demonstrated an improved capacity to inhibit and displace pathogens from human mucus than were human strains, further reporting that bifidobacteria were capable of producing antimicrobial peptides directed against Helicobacter pylori (29). Cross-species utilization of probiotics may therefore provide another important level of complexity for probiotic utility in clinical disorders such as IBD. In certain allergic disorders, including atopic eczema, probiotic influences on mucosal barrier function may be operative (30), whilst multiple mechanisms may account for anti-neoplastic effects (31). Probiotics, tolerance and the immune responseSince control of bowel inflammation has generally been recognised as paramount in IBD, it is perhaps not surprising that down-regulation of mucosal inflammation has been identified as the primary focus of both conventional and probiotic-targeted therapy, fuelled by the cumulative clinical experience with anti-TNF based treatment successes (32). Rodents and humans are normally tolerant to autologous microbiota, and an association between breakdown of this tolerance and the development of chronic intestinal inflammation has been demonstrated (32). Potentially, pathological responses to components of the intestinal flora may occur under normal physiological conditions, but these may be suppressed by immunoregulatory mechanisms. In a recent review by Thompson-Chagoyán et al (33), 11 models of IBD have been described, in which inflammation was found to be dependent on the presence of normal flora; the absence of normal flora being associated with non-appearance of the condition (34,35,36,37). This phenomenon has been reported across species (mice, rats and guinea pigs), and to occur in manipulated organisms such as transgenic mice with targeted deletion of the T-cell receptor (TCRα), that spontaneously develop colitis in response to the gut microbiota (38). Mucosal inflammation in rats and mice with induced IBD has also been reported to respond to treatment with broad-spectrum antibiotics (39). Moreover; in some animal model systems, colonisation with normal flora results in the rapid development of T-cell-mediated gut inflammation which can be transferred to other animals using activated T cells directed against enteric bacteria (40). Nevertheless, it appears that not all commensal bacteria have an equivalent ability to induce mucosal inflammation, which is also influenced by the host genetic background.Following cell surface recognition via Toll-like receptors (41), the anti-inflammatory effects of probiotics require signalling with the epithelium and hence, the mucosal immune system (42). Although transduction of bacterial signals into host immune responses presumably involves more than one pathway, NF-kappaB has been established as a central regulator of epithelial responses to invasive pathogens (43,44). Non-pathogenic components of the flora may attenuate pro-inflammatory responses by delaying degradation of IkappaB, the counter-regulatory factor to NF-kappaB (44). Indeed, it may be that probiotic bifidobacteria and lactobacilli may not use the same mechanism to achieve their anti-inflammatory effects as other signal transduction pathways begin to emerge. Clearly, a better understanding of the interplay between prokaryotes and eukaryotes, and subsequent effects on metabolic activity, will facilitate our knowledge of probiotic mechanisms. Bacterial adjuvants, including peptidoglycan, lipopolysaccharide, and DNA (CpG) bind to membrane-bound Toll-like receptors (TLR-2, 4, and 9. respectively), or cytoplasmic (NOD1 and NOD2) receptors (pattern recognition receptors), that activate nuclear factor-kappaB and transcription of many proinflammatory cytokines (45). Prokaryotic DNA perhaps represents the first description of a growing number of bacteria-sourced factors with the potential to alter host epithelial and mucosal immune responses. Un-methylated cytosine–guanine (CpG)-containing DNA, the ligand for Toll-like receptor 9 (TLR9), is a recently recognized microbial product with immuno-stimulatory and immuno-regulatory effects (46). TLR9 is expressed by many cell types located in the intestine, including epithelial cells and dendritic cells, and subcutaneous administration of immuno-stimulatory DNA has been reported to reduce the severity of experimental and spontaneous colitis in murine models of IBD (47) via a mechanism attributed to TLR9 signalling (48).A decrease in the secretion of pro-inflammatory cytokines, IFN-gamma, TNF-alpha and IL-12, and interference with bacterial adherence to the epithelium has been demonstrated following probiotic administration, associated with NF-kappaB inhibition, heat-shock protein induction and proteasome inhibition, although NF-kappaB induction has also been demonstrated (49,50). Unexpectedly, many of these beneficial effects have been achieved not only by live bacteria, but also by gamma-irradiated non-viable bacteria, bacterial DNA components and probiotic-cultured media (49,51). Investigations into probiotic supernatants and their therapeutic potential in IBD are therefore forming the basis for new directions in IBD research. In summary, although mechanisms of probiotic action may vary, depending on the experimental or clinical context, and depending on differences in the host and in the bacterial strain, the engagement with host immunity is pivotal to probiotic action in IBD. Probiotics as therapeutic agents in IBDThe human colon is a densely populated microbial ecosystem with several hundred bacterial species usually present with a total weight estimated to be several hundred grams (52). There are up to 1013–1014 total bacteria in the human intestinal tract, representing 10- to 20-fold more than the total number of tissue cells in the entire body, with most bacteria being obligate anaerobes, including clostridia, eubacteria, bacteroides groups and the genus bifidobacterium, such as Bifidobacterium bifidum and Bifidobacterium infantis (53).Probiotic efficacy has been confirmed in animal model studies by several investigators with different probiotic strains (37,53), although the use of probiotic therapy for IBD has only recently attracted serious interest from clinicians. Clinical trials in Crohn's disease with organisms, including lactobacillus (54) yeast (13), and coliforms (56), have been confounded by small patient numbers, differences in disease activity and variations in disease distribution (56). These deficiencies have been exacerbated by a growing, but poorly regulated, commercial market for probiotics, often linked with somewhat tenuous, or exaggerated, claims for health benefits. Historically, the stability, optimal dose-range, frequency of administration and vehicle for delivery, have rarely been determined. However, at present in Europe, well-designed and appropriately controlled trials of individual probiotic preparations are underway in both Crohn's disease and ulcerative colitis, to ascertain their efficacy in both active disease and in prevention of relapse.Individual probiotics and IBD treatmentThe predominant, potentially health-enhancing, bacteria in IBD are the bifidobacteria and lactobacilli, both of which belong to the lactic acid bacteria (LAB) group (57). These two genera do not include any significant pathogenic species and their dominance in the faeces of breast-fed babies is thought to impart protection against infection (58,59). Most commercially available probiotics meet minimum selection criteria including acid and bile resistance and survival during gastrointestinal transit, but an ideal individual probiotic strain for any given indication has still yet to be defined. Lactobacilli and bifidobacteria have traditionally been the most common candidates for probiotic-based treatments in IBD. Although there is a growing body of scientific literature and a wealth of anecdotal information supporting the utility of lactobacillus strains as therapeutic agents in a range of alimentary disorders, progress has perhaps been hampered by poorly-controlled associations between probiotic administration and clearly-defined clinical end-points. Recently, Hawrelak et al (60) conducted a systematic review of six clinical trials investigating the capacity for Lactobacillus rhamnosus GG to prevent the onset of antibiotic-associated diarrhoea. As data sources, these investigators employed computer-based searches of MEDLINE, CINAHL, AMED, the Cochrane Controlled Trials Register and the Cochrane Database of Systematic Reviews and the bibliographies of relevant papers and previous meta-analyses. Four of the six trials found a significant reduction in the risk of antibiotic-associated diarrhoea with co-administration of Lactobacillus GG; one trial reported a reduced number of days with antibiotic-induced diarrhoea, whilst the final trial found no benefit of Lactobacillus GG supplementation. These results are not unique in the context of retrospective probiotic studies, highlighting the need for additional research and the development of strictly-controlled predictive systems to clarify the effectiveness of Lactobacillus-based treatments beyond the effects of LGG in prevention of antibiotic-associated diarrhoea.Bifidobacterium, a member of the dominant microbiota (i.e. >108–109 colony forming unit (CFU)/g, represent up to 25% of the cultivable faecal bacteria in adults and 80% in infants. As probiotic agents, bifidobacteria have been studied for their efficacy in the prevention and treatment of a broad spectrum of animal and/or human gastrointestinal disorders, such as colonic transit disorders, intestinal infections, and colonic adenomas and cancer (10). Indeed, certain strains (eg Bifidobacterium animalis strain DN-173 010) have been reported to prevent or alleviate infectious diarrhoea through effects on the immune system and resistance to colonization by pathogens and there is some evidence that certain bifidobacteria may actually protect the host from pro-carcinogenic activity of intestinal flora (10).It should be noted, however, that probiotic properties have not been attributed solely to bacterial sources, since non-bacterial organisms, such as Saccharomyces boulardii, or even nematode parasites have been utilised for probiotic purposes (13,65) In summary, it seems unlikely that a single probiotic will be equally suited to all indications; selection of strains for disease-specific indications will be required, and it is in this capacity that well-conducted animal model studies will prove a valuable tool ConclusionsProbiotics continue to hold great promise for IBD treatment and prevention, but despite some significant advances, it would be fair to say that this field of research is still in its infancy. Understanding the mechanisms responsible for the beneficial effect of probiotics in inflammatory bowel disease and experimental colitis will aid our understanding of the role of endogenous and exogenous bacteria in IBD aetiology and pathogenesis. The finding that live probiotics may not be essential for therapeutic effects, and that these effects may also be obtained by the systemic route of administration, could have a major impact on the use and manufacture of probiotics. Phenotypic and genomic characterisation of probiotic strains will be required, together with clarification of their mechanisms of action across a broad range of rigorously-controlled clinical settings. Moreover, results of probiotic studies demonstrating efficacy in a given clinical setting should not be extrapolated to other disease indications without separate controlled assessments. Over the past decade, there have been quantum advances in probiotic-based research in IBD. Initial, largely anecdotal, reports of lactobacillus efficacy have been replaced by the development of specific ‘designer’ probiotics, which together with identified prebiotics, could result in a significant repertoire of designer synbiotics for IBD treatment. Such an array of treatment options, combined with genetic advances in IBD susceptibility and probiotic responsiveness, could eventually result in the exciting emergence of individually-tailored probiotic-sourced formulations. The major challenge in the short-term will be the development of rigorous in vivo and in vitro testing systems to rapidly determine the suitability of any given probiotic-based formulation, not only to a specific condition, but also to the individual.